0% found this document useful (0 votes)
50 views115 pages

4 - Bioavailability and Bioequivalence

The document discusses bioavailability (BA) and bioequivalence (BE) of drug products, emphasizing their significance in ensuring therapeutic efficacy. It outlines the definitions, factors affecting BA, and methods for evaluating BA and BE, including pharmacokinetic studies and clinical trials. Additionally, it highlights the importance of demonstrating that generic drugs are bioequivalent to brand-name counterparts to ensure safety and effectiveness.

Uploaded by

Misgana Getachew
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
50 views115 pages

4 - Bioavailability and Bioequivalence

The document discusses bioavailability (BA) and bioequivalence (BE) of drug products, emphasizing their significance in ensuring therapeutic efficacy. It outlines the definitions, factors affecting BA, and methods for evaluating BA and BE, including pharmacokinetic studies and clinical trials. Additionally, it highlights the importance of demonstrating that generic drugs are bioequivalent to brand-name counterparts to ensure safety and effectiveness.

Uploaded by

Misgana Getachew
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 115

4.

Bioavailability and Bioequivalence



• For drugs administered extravascularly and intended for systemic
effects, absorption is a prerequisite for therapeutic activity.

o Factors affecting drug absorption & BA have been discussed


previously.

o The main objective here will be to discuss:

• concepts of BA and BE

• general methods to evaluate BA and BE.

• scientific principles & regulatory perspectives related to BA & BE

2

• Single-source drug products ??

o Drug products for which the patent has not yet expired,

o Has certain exclusivities so that only one manufacturer can


make it.

o Are usually brand-name (innovator) drug products.

o The patent and other exclusivities for the brand-name drug


expires after a certain period of time.

3
Drug product selection and
generic drug product
… substitution are major
responsibilities for a
• Multisource drug product ?? pharmacist

o Drug product that contains the same API in the same dosage
form and is marketed by different manufacturers.

o The formulation and method of manufacture may be different,

• This can affect the bioavailability and stability of the drug

o The generic drug manufacturer must demonstrate that the


generic drug product is bioequivalent and therapeutically
equivalent to the brand-name drug product.

4

Definition of Terms

• Bioavailability

o The rate and extent to which the API is absorbed from a drug
product and becomes available at the site of action.

• Equivalence

o Relationship in terms of bioavailability, therapeutic response, or a


set of established standards of one drug product to another.

5

• Bioequivalent drug products

o Pharmaceutical equivalent or pharmaceutical alternative


products that display comparable bioavailability.

o Showing the same rate and extent of absorption.

o Bioequivalent drug products may contain different inactive


ingredients provided the differences do not affect the safety or
efficacy of the product.

6

• Brand name

o The trade name of the drug.

o This name is privately owned by the manufacturer.

• Chemical name

o The name used to indicate the chemical structure of the drug

7

• Generic name

o The established, nonproprietary, or common name of the API,

o Was initiated in 1950 by WHO,

o Aim: to provide health professionals with a unique and


universally available designated name to identify each
pharmaceutical substance.

8

o Drugs from the same therapeutic or chemical class are usually
given names with the same stem,

o Examples:

• -ase for enzymes (e.g. alteplase)

• -azepam for benzodiazepines (e.g. diazepam and oxazepam)

• -pril for ACE inhibitors (e.g. captopril)

9

• Drug product

o The finished dosage form

• Drug substance

o A drug substance is the active pharmaceutical ingredient (API)

10

• Generic substitution

o The process of dispensing a different brand or an unbranded


drug product in place of the prescribed drug product.

o The substituted drug product contains the same API or


therapeutic moiety as the same salt or ester in the same dosage
form

11

• Pharmaceutical alternatives

o Drug products that contain the same therapeutic moiety but as


different salts, esters, or complexes.

o Different dosage forms and strengths within a product line by a


single manufacturer

o E.g. A tablet and capsule containing the same active ingredient


in the same dosage strength.

12

• Pharmaceutical equivalents

o Drug products in identical dosage forms that contain the same


API/same therapeutic moiety,

o Use the same route of administration,

o These may differ in characteristics such as shape, scoring


configuration, release mechanisms, packaging, excipients
(including colors, flavors, preservatives), expiration time, and,
within certain limits, labeling.

o Must meet the same content uniformity, disintegration times,


and/or dissolution rates.

13

• Pharmaceutical substitution

o The process of dispensing a pharmaceutical alternative for the


prescribed drug product.

o E.g., ampicillin suspension is dispensed in place of ampicillin


capsules, or tetracycline HCl is dispensed in place of tetracycline
phosphate.

14

• Therapeutic alternatives

o Drug products containing different APIs that are indicated for the
same therapeutic or clinical objectives.

• Therapeutic equivalents

o Drug products are considered to be therapeutic equivalents only


if they are pharmaceutical equivalents and if they can be
expected to have the same clinical effect and safety profile

15

• Therapeutic equivalent drug products must be:

• Approved as safe and effective

• Pharmaceutical equivalents

• Bioequivalent

• Adequately labeled

• Manufactured in compliance with CGMP regulations

16

• Therapeutic substitution

o The process of dispensing a therapeutic alternative in place of


the prescribed drug product.

17
Bioavailability
• Bioavailability is defined as the rate and extent of drug
absorption.

o The fraction of an administered dose of the drug that reaches


the systemic circulation unchanged,

o The assumption here is that the intact drug is the


therapeutically active form.

o This definition would not be valid in the case of prodrugs.

18

• The issue of Bioavailability and Bioequivalence has become a
critical issue in pharmacy.

o Increase in the use of generic drug products (lowering health


care costs).

o Much of the prescription drugs are available from more than one
source.

o The availability of different formulations of the same API given


at the same strength and in the same dosage form.

o These calls for selecting those that are therapeutically


equivalent.
19

• Bioavailability studies are used to define the effect of changes


in the physicochemical properties of the drug and the effect of
the drug product (dosage form) on the PK of the drug.

• Bioavailability studies are performed for both approved API


and for new drugs for marketing

20

• For new drugs

1) To establish essential PK parameters including:

• Rate and extent of systemic absorption

• Rates of excretion and metabolism

• Elimination half-life

21

2) To establish dosage regimens:

• Dose, Frequency of administration

• Treatment duration

22

3) To determine influence of:

• Excipients

• Manufacturing procedures

• Patient related factors on biological performance of new drug


formulation

23

• For approved drugs

o To develop new DF or to improve on existing DF.

o To determine the bioavailability and to characterize the PK of the


new formulation, new dosage form, or new salt or ester relative
to a reference formulation.

24

• In approving a drug product for marketing, it must be


ensured that the drug product is safe and effective for its
labeled indications for use.

o The drug product must meet all applicable standards of identity,


strength, quality, and purity.

25

Significance of Bioavailability

• Drugs having low therapeutic index

o E.g. Cardiac Glycosides, Quinidine, Phenytoin etc.

• Drugs whose peak levels are required for the effect of drugs

o E.g. Phenytoin, Phenobarbitone, Primidone, Sodium Valporate,


Anti-hypertensives, Antidiabetics and Antibiotics.

26

• Drugs that are absorbed by an active transport

o E.g. Amino acid analogues, Purine analogues etc.

• Drugs which are disintegrated in the alimentary canal and


liver

o E.g. Chlorpromazine or those which under go first pass


metabolism.

27

• Formulations that give sustained release of drug

• Formulations with smaller disintegration time than dissolution


rate

• In addition, any new formulation has to be tested for its


bioavailability profile

28

Factors Affecting Bioavailability

• For a drug which is administered orally to be 100%


bioavailable, the entire dose must move from the dosage form
to the systemic circulation.

• The drug must therefore be:

o Completely released from the dosage form

o Fully dissolved in the GI fluids

29

o Stable in solution in the GI fluids

o Pass through the GI barrier into the mesenteric circulation


without being metabolized

o Pass through the liver into the systemic circulation unchanged.

• Anything which affects either of these will influence the


bioavailability.

30

• Three distinct factors are involved to influencing


bioavailability:

1. Pharmaceutical factors

2. Patient related factors

3. Route of administration

31

• Pharmaceutical factors

A. Physicochemical properties of the drug: Particle size, Crystalline


structure, Salt form

B. Formulation and manufacturing variables: Disintegration and


dissolution time, Pharmaceutical ingredients, Special coatings,
Nature and type of dosage form

32

• Patient related factors

A. Physiologic factors

• Variations in pH of GI fluids • Pre-systemic and


first-pass metabolism
• Gastric emptying rate
• Age, sex
• Intestinal motility
• Disease states

33

B. Interactions with other substances:

• Food

• Fluid volume

• Other drugs

34

• Route of administration

o Parenteral administration

o Oral administration

o Rectal administration

o Topical administration

35

Absolute and Relative Bioavailability

• Drug concentrations usually cannot be readily measured


directly at the site of action.

• Most bioavailability studies involve the determination of drug


concentration in the blood or urine.

• It is possible to obtain an indirect measure of drug response


by monitoring drug levels in the blood or urine.

36

• The fraction of the dose that actually reaches the bloodstream


is important since this represents the "effective dose" of a
drug.

o This is generally less than the amount of drug actually


administered in the dosage form.

37

• Absolute bioavailability (F) is the fraction of an


administered dose which actually reaches the systemic
circulation.

• ‘‘F’’ ranges from F = 0 (no drug absorption) to F = 1


(complete drug absorption).

• AUC (total amount of unchanged drug that reaches the


systemic circulation) is used to calculate F.

38

• F is determined by comparing the respective AUCs of the test


product and the same dose of drug administered
intravenously.

• Knowledge of F is needed to determine an appropriate oral


dose of a drug relative to an IV dose.

39

• Relative / Comparative bioavailability: refers to the
availability of a drug product as compared to another product
of the same drug given in the same dose.

o It determines the effects of formulation differences on drug


absorption.

o The relative bioavailability of two products A and B can be


obtained by comparing their respective AUCs.

40

Methods of Assessing Bioavailability

• Bioavailability determinations are performed:

o To ensure that a given drug product will get the therapeutic


agent to its site of action in an adequate concentration.

o To compare the availability of a drug substance from different


dosage forms or from the same dosage form produced by
different manufacturers.

41

• Different methods are used to assess drug bioavailability

o Pharmacokinetic Studies

o Pharmacologic Effect (Pharmacodynamic) Studies

o Comparative Clinical Trials

o In Vitro Dissolution Studies

42

• Pharmacokinetic Studies

o Emphasizes the release of the drug substance from the drug


product with absorption into the systemic circulation.

o Assumption: drug concentration at the site of action is


proportional to that in the systemic circulation.

o Pharmacokinetic studies use:

• Plasma drug concentration data

• Urinary drug excretion data

43

• Pharmacologic effect (Pharmacodynamic) Studies

o Establishment of a dose-response relationship.

• Comparative Clinical Trials

o In the absence of PK and PD approaches, adequate and well-


controlled clinical trials may be used to establish BA.

44

• In Vitro Dissolution Studies

o Measures the rate and extent of dissolution of the drug in an


aqueous medium in the presence of one or more excipients
contained in the drug product.

o A potential bioavailability problem may be uncovered by a


suitable dissolution method.

o The optimum dissolution testing conditions differ with each drug


formulation.

45

• BA and BE measures are frequently expressed in systemic exposure
measures.

• These measures of systemic exposure are assumed to relate in


some way to safety and efficacy outcomes.

• These may also provide additional useful information about

• ADME of the drug

• Dose proportionality

• Nutrients effects on drug absorption

• Performance of the formulation

46

PK Studies

• One method for assessing the bioavailability of a drug product


is through the demonstration of a clinically significant effect.

• However, this is complex, expensive, time-consuming and


require a sensitive and quantitative measure of the desired
response.

• Further, response is often quite variable, requiring a large test


population.

47

• Quantification of pharmacologic effect is another possible way


to assess a drug’s bioavailability.

• This method is based on the assumption that a given intensity


of response is associated with a particular drug concentration
at the site of action.

48

• However,

o Monitoring of pharmacologic data is often difficult

o Precision and reproducibility are difficult to establish, and

o There are only a limited number of pharmacologic effects (e.g.


heart rate, body temperature, blood sugar levels) that are
applicable to this method.

49

• The current method to assess the clinical performance of a


drug involves measurement of the drug concentrations in the
blood or urine.

o A single dose of the drug product is administered

o Blood and/or urine samples are collected over a period of time

o Based on the blood concentration as a function of time and/or


urinary excretion profile, inferences are drawn regarding the rate
and extent of absorption of the drug.

50

Blood level studies

• The most common type of bioavailability studies.

• Assumption: there is a direct relationship between the


concentration of drug in blood/plasma and the concentration
of drug at the site of action.

o By monitoring the concentration in the blood, it is thus possible


to obtain an indirect measure of drug response.

51

• The key parameters to note from the plasma drug


concentration data are:

o AUC - The area under the plasma concentration-time curve

• The AUC is proportional to the total amount of drug reaching


the systemic circulation.

• It characterizes the extent of absorption.

o For many drugs, the AUC is directly proportional to the


administered dose.

52

• AUC is area under plasma level–time curve from t=0 to t=∞

o Where, ‘t’ is last time point of plasma sample collection

• is calculated by trapezoidal rule:

= (1/2) (C1+C2) (t2-t1) + (1/2) (C2+C3) (t3-t2) + … +


(1/2) (Cn-1+Cn) (tn-tn-1)

53

54

o Cmax - The maximum drug concentration.

• It is a function of both the rate and extent of absorption.

• Cmax will increase with an increase in the dose, as well as


with an increase in the absorption rate.

o tmax - The time at which the Cmax occurs.

• It reflects the rate of drug absorption, and

• tmax decreases as the absorption rate increases.

55
56

• If all other factors are constant, such as the extent of


absorption and rate of elimination:

o Cmax is proportional to the rate of absorption.

o tmax is inversely proportional to the absorption rate.

• Thus, the faster the absorption of a drug the higher the Cmax
will be and the less time it will take to reach the maximum
concentration.

57

• Cmax could provide:

o Indications that drug is sufficiently systemically absorbed to


provide therapeutic response

o Warning of possibly toxic levels of drug

58
59

• At tmax:

o Peak drug absorption occurs (i.e. Cmax is obtained)

o Rate of absorption exactly equals rate of drug elimination

o Drug absorption still continues after tmax is reached, but at


slower rate

60

• Note:

o Cmax and tmax are measures of rate of absorption.

o AUC is measure of extent of absorption.

61

Urinary Excretion Data

• Measures the cumulative amount of unchanged drug excreted


in the urine.

• Involve collection of urine samples and the determination of


the total quantity of drug excreted in the urine as a function
of time.

62

• Assumption: Urinary excretion of the unchanged drug is


directly proportional to the plasma concentration of total drug.

o The total quantity of drug excreted in the urine is a reflection of


the quantity of drug absorbed from the GIT.

• This technique of studying bioavailability is most useful for


those drugs that are not extensively metabolized prior to
urinary elimination.

63

• As a rule-of-thumb, determination of BA using urinary


excretion data should be conducted only if at least 20% of a
dose is excreted unchanged in the urine after an IV dose.

• In addition:

o The fraction of drug entering the bloodstream and being


excreted intact by the kidneys must remain constant.

o Collection of the urine has to continue until all the drug has been
completely excreted

64

• The cumulative amount of drug excreted in the urine, 𝑫∞𝒖 , is


related directly to the total amount of drug absorbed.

• Experimentally,

o Urine samples are collected periodically after administration of a


drug product.

o Each urine specimen is analyzed for free drug using a specific


assay.

o A graph is constructed that relates the cumulative drug excreted


to the collection-time interval

65

Fig. The relationship between the cumulative amount of drug


excreted in the urine and the plasma level–time curve
• When the drug is almost completely eliminated (point C), the
plasma concentration approaches zero and the maximum
amount of drug excreted in the urine, 𝑫∞
𝒖 , is obtained.

66

• Important parameters from urinary excretion data:

𝒅𝑫𝒖
o (The rate of drug excretion)
𝒅𝒕

• Because most drugs are eliminated by a first-order rate


process,

o the rate of drug excretion is dependent on the first-order


elimination rate constant k and the concentration of drug
in the plasma.

67

Fig. The relationship between the plasma level–time curve and


the rate of urinary drug excretion
• The maximum rate of drug excretion is at point B, whereas the
minimum rate of drug excretion is at points A and C.

68

o 𝒕∞ (The total time for the drug to be excreted)

• From the above graph (B), point C is related to the total time
required after drug administration for the drug to be
absorbed and completely excreted t = ∞.

• The 𝒕∞ is a useful parameter in bioequivalence studies that


compare several drug products.

69

• Urinary excretion data are used:

o Primarily for assessing extent of drug absorption.

o To estimate the rate of absorption.

• These are subject to a high degree of variability

o Urinary excretion of drug is not recommended as a substitute for


blood concentration data

70

Single-dose versus Multiple-Dose

• Most bioavailability evaluations are made on the basis of


single-dose administration.

• Single doses are not representative of the actual clinical


situation.

• In most instances, patients require repeated administration of


a drug.

71

• When a drug is administered repeatedly at fixed intervals,
with the dosing frequency less than five half-lives, drug will
accumulate in the body and eventually reach a plateau, or a
steady-state.

• At steady-state, the amount of drug eliminated from the body


during one dosing interval is equal to the available dose (rate
in = rate out).

• Therefore, the AUC during a dosing interval at steady-state is


equal to the total AUC obtained when a single dose is
administered.

72

• Multiple dose vs. single dose studies in bioavailability studies:

Advantages

o Eliminates the need to extrapolate the plasma concentration


profiles to obtain the total AUC after a single dose

o Eliminates the need for a long washout period between doses

73

o More closely reflects the actual clinical use of the drug

o Allows blood levels to be measured at the same concentrations


encountered therapeutically

o Because blood levels tend to be higher than in the single-dose


method, quantitative determination is easier and more reliable

o Saturable PK, if present, can be more readily detected at steady-


state

74

Limitations

o Requires more time to complete

o More difficult and costly to conduct

o Greater problems with compliance control

o Greater exposure of subjects to the test drug, increasing the


potential for adverse reactions

75

• In general, when a drug obeys linear, first-order kinetics,

o It is possible to estimate the results that would be obtained


during multiple dosing from single-dose studies.

o Projection is easily made with regard to the extent of absorption,


using the AUC following a single dose.

76

• In the case of drugs exhibiting nonlinear kinetics,

o Establishing a linear relationship between single- and multiple-


dose bioavailability data has proven to be a difficult task.

o Thus, it has been recommended that for drugs with either


saturable elimination or a nonlinear first-pass effect, steady-state
studies be carried out to assess their bioavailability

77
Bioequivalence
• BE refers to pharmaceutically equivalent drug products where
the rates/extents of bioavailability of the actives are not
significantly different under suitable test conditions.

• In other words, this is a comparison of two or more products


with respect to their bioavailability.

• BE means the absence of a greater-than-allowable difference


between the systemic bioavailability of a test product and that
of a reference product.

78

• BE cannot be claimed based on in-vitro testing only or on the


basis of animal studies only.

• BE of human drugs must be determined in humans via


established measures of bioavailability.

Once BE has been established in a statistically significant


manner subsequent batches of the same product are
deemed bioequivalent based on in-vitro measures.

79

• The basis for BE study is that if two formulations exhibit


similar drug concentration-time profiles in the blood/plasma,
they should exhibit similar therapeutic effects.

• BE studies provide a quality control tool to monitor production


and manufacturing changes.

80

• Three situations have been defined in which BE studies are


required:

1) When the proposed marketed dosage form is different from


that used in pivotal clinical trials,

2) When significant changes are made in the manufacture of the


marketed formulation, and

3) When a new generic formulation is tested against the


innovator’s marketed product.

81

• Bioequivalent drug products that have the same systemic
drug bioavailability will have the same predictable drug
response.

• Differences in the predicted clinical response or an adverse


event may occur due to:

o Differences in the PK and/or PD behavior of the drug among


individuals or

o Differences in the bioavailability of the drug from the drug


product.

82

• Variable clinical responses among individuals that are


unrelated to bioavailability may be due to differences in the
PD of the drug.

o May be due to differences in receptor sensitivity to the drug.

o May be related to age, drug tolerance, drug interactions, and


unknown pathophysiologic factors.

83

• In a few cases, a drug product that differs from the reference


listed drug in its rate of absorption, but not in its extent of
absorption, may be considered bioequivalent

o If the difference in the rate of absorption is intentional and


and/or the rate of absorption is not detrimental to the safety and
effectiveness of the drug product.

84

• Some biopharmaceutical properties of the API or the


formulation which will be associated with variable
bioavailability and/or a bioequivalence problem:

o The API has low solubility in water.

o The dissolution rate of the products is slow.

o The particle size and/or surface area of the API is critical in


determining its bioavailability.

85

o Certain structural forms of the API dissolve poorly, thus affecting


absorption.

o Drug products that have a high ratio of excipients to active


ingredients

o Specific inactive ingredients either may be required for


absorption of the API or therapeutic moiety or may interfere
with such absorption.

o The API is absorbed in large part in a particular segment of the


GI tract or is absorbed from a localized site.

86

o The degree of absorption of the API is poor.

o The therapeutic moiety is rapidly metabolized or excreted, so


that rapid dissolution and absorption are required for
effectiveness.

o The API is unstable in the GIT and requires special coatings or


formulations to ensure adequate absorption.

o The drug product is subject to dose-dependent kinetics, and the


rate and extent of absorption are important to bioequivalence.

87

Design of Bioequivalence Studies

• BE studies are performed to compare the bioavailability of the


generic drug product to the brand-name product.

• Statistical techniques should be of sufficient sensitivity to


detect differences in rate and extent of absorption.

88

• The basic design for a bioequivalence study is determined by:

o The scientific questions to be answered,

o The nature of the reference material and the dosage form to be


tested,

o The availability of analytical methods,

o Benefit–risk and ethical considerations with regard to testing in


humans.

89

• Generally, the study is performed in normal, healthy male and


female volunteers who have given informed consent to be in
the study.

• Critically ill patients are not included unless the attending


physician determines that there is a potential benefit to the
patient.

• The number of subjects in the study will depend on the


expected intersubject and intrasubject variability.

90

• Three different studies may be required:

1. Fasting Study

• BE studies are usually evaluated by a single-dose, two-


period, two-treatment, two-sequence, open-label,
randomized crossover design comparing equal doses of
the test and reference products in fasted, adult, healthy
subjects.

• This study is required for all immediate-release and modified-


release oral dosage forms.
91

• The subjects should be in the fasting state (overnight fast of
at least 10 hours) before drug administration,

• Should continue to fast for up to 4 hours after dosing,

• No other medication is normally given to the subject for at


least 1 week prior to the study.

• Single dose of the drug is given in randomized crossover


design,

• Blood sampling, processing, and adequate description of the


plasma drug concentration–time profile …?

92

2. Food Intervention Study

• Co-administration of food with an oral drug product may


affect the bioavailability of the drug.

• Food intervention studies are generally conducted using meal


conditions.

• The test meal is a high-fat (approximately 50% of total


caloric content of the meal) and high-calorie
(approximately 800–1000 calories) meal.

93

• A typical test meal is two eggs fried in butter, two strips of
bacon, two slices of toast with butter, 4 ounces (~28g) of
brown potatoes, and 8 ounces of milk (~1 cup).

• Following an overnight fast of at least 10 hours, subjects are


given the recommended meal 30 minutes before dosing.

• The meal is consumed over 30 minutes, with administration


of the drug product immediately after the meal.

• Single dose of the drug is given in randomized crossover


design,

• Blood sampling, processing, and adequate description of the


plasma drug concentration–time profile …?
94

3. Multiple-Dose (Steady-State) Study

• For these studies, three consecutive trough concentrations


(Cmin) on three consecutive days should be determined to
ascertain that the subjects are at steady state.

• The last morning dose is given to the subject after an


overnight fast, with continual fasting for at least 2 hours
following dose administration.

• Blood sampling is performed similarly to the single-dose


study.

95

• BE study should be designed in such a way that the


formulation effect can be distinguished from other effects

o Parallel Design

o Cross-over Design

96

Parallel Design

o Two formulations are administered (randomly) to two groups of


volunteers.

o It do not address inter-subject variations.

o Note: most of the time, inter-subject variation is greater than


variation between formulations.

97

o Each subject administered test or the reference product.

o Between-subject comparison.

o Only recommended for extremely long half-life drugs.

98

Cross-over Design

o Subjects who meet the inclusion and exclusion study criteria and
have given informed consent are selected at random.

o A complete crossover design is usually employed.

99

o Each subject administered both test and the reference products

• Within-subject comparison.

• Inter-subject variables such as age, weight, differences in


metabolism, etc., are minimized.

• Each subject acts as his/her` own control.

o Preferred

100

• Types of crossover design:

• Latin square/Complete crossover design

• Balanced incomplete block design (BIBD)

• Replicated crossover design

101

Latin-square crossover design

o Each subject receives each drug product only once, with


adequate time between medications for the elimination of the
drug from the body (washout period).

o E.g. Complete cross-over study of two products A and B in 12


subjects;

102

Advantages

o Minimizes inter-subject variability in study

o Requires less number of subjects to get meaningful results

o Minimizes carryover effect

• All patients don’t receive same drug product on same day


and in same order

• Variations due to sequence and period are reduced

103

Disadvantages

o Requires longer time to complete since washout period exists

• Longer biological half-life

o Time to complete trial depends on number of formulations


evaluated in study

o Increased number of study periods leads to high subject


dropouts

o Medical ethics doesn’t allow too many trials on a subject


continuously for longer time

104

Balanced incomplete block design (BIBD)

o Each subject receives not more than two formulations

o Each formulation is administered same number of times

o Each pair of formulation occurs together in same number of


subjects

105

o E.g. BIBD for four formulations A, B, C, D in 12 subjects

106

Replicated cross-over design

o Used for drug product containing drug that has high intra-
subject variability

o Generally, four-period, two-sequence, two-formulation design is


recommended.

107

In Vitro - in Vivo Correlations

• There are many satisfactory correlations between absorption


parameters and in vitro dissolution tests.

• There have also been cases where a drug has failed to meet
compendia dissolution standards but has demonstrated
adequate BA.

• Even if an in vitro test could be designed that would


accurately reflect the dissolution process in the GIT,
dissolution is only one of many factors that affect a drug’s BA.

108

• Dissolution studies would not predict poor bioavailability due


to:

• Presystemic metabolism

• Instability in gastric fluid

• Complexation with another drug or food component

109

• The ultimate evaluation of a drug product’s performance
under the conditions expected in clinical therapy must be an
in-vivo test

• A dissolution test is unlikely to entirely replace bioavailability


testing.

• In-vitro methods are important in the development and


optimization of dosage forms while in-vivo tests are essential
in obtaining information on the behavior of medication in
living organisms.

110

Bio waivers

• Waivers of in-vivo bioequivalence studies.

• Human in-vivo test is considered to be preferable to other


approaches for the most accurate determination of BE.

• In some cases, in-vitro dissolution testing may be used in lieu


of in-vivo BE studies.

111

• When there is a strong correlation b/n dissolution of the drug
and the BA of the drug,

• The in vitro comparative dissolution tests could be


sufficient to demonstrate bioequivalence.

o E.g. When the drug product is in the same dosage form but in
different strengths,

• an in-vivo bioequivalence study of the lower strengths can be


waived based on the dissolution tests and an in-vivo
bioequivalence study on the highest strength.

112

• Criteria:

o The drug product is a solution (in the same solvent and


concentration) intended solely for IV administration.

o The drug product is a topically applied preparation intended for


local therapeutic effect.

o The drug product is an oral dosage form that is not intended to


be absorbed, e.g., an antacid.

113

o The drug product is administered by inhalation and contains the


API in the same dosage form as a drug product that is the
subject of an approved,

o The drug product is an oral solution that contains an API in the


same concentration as a drug product that is approved and
contains no inactive ingredient that is known to significantly
affect absorption of the API.

114

o Reading assignment –

• Biopharmaceutical classification Scheme/System (BCS)

115

You might also like